151
|
Minamino T, Ito Y, Ohkubo H, Shimuzu Y, Kojo K, Nishizwa N, Amano H, Narumiya S, Koizumi W, Majima M. Adhesion of platelets through thromboxane A₂ receptor signaling facilitates liver repair during acute chemical-induced hepatotoxicity. Life Sci 2015; 132:85-92. [PMID: 25921763 DOI: 10.1016/j.lfs.2015.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/02/2015] [Accepted: 03/13/2015] [Indexed: 01/18/2023]
Abstract
AIMS Platelets have been suggested to play an important role in liver regeneration and repair after hepatic resection and acute liver injury. However, the underlying mechanisms of liver repair remain elusive. Signaling through thromboxane prostanoid (TP) receptor participates in inflammation and tissue injury through platelet aggregation. On the other hand, TP receptor signaling also is involved in tissue repair and tumor growth through angiogenesis. The present study was examined whether or not TP receptor signaling contributes to liver repair and sinusoidal restoration from acute liver injury through platelet adhesion to the hepatic sinusoids. MAIN METHODS Carbon tetrachrolide (CCl4) was used to induce acute liver injury in TP receptor knockout mice (TP(-/-) mice) and their wild-type littermates (WT mice). KEY FINDINGS Compared with WT mice, TP(-/-) mice exhibited delayed in liver repair and sinusoidal restoration after CCl4 treatment, which were associated with attenuated hepatic expression of pro-angiogenic factors. Intravital microscopic observation revealed that adhering platelets to the sinusoids was increased in WT livers during the repair phase as compared with TP(-/-) livers, and platelet adhesion was dependent on TP receptor signaling. The levels of hepatocyte growth factor (HGF) in platelets from WT mice treated with CCl4 for 48h were greater than those form TP(-/-) mice, and HGF enhanced the expression of angiogenic factors in cultured human umbilical vein endothelial cells (HUVECs). SIGNIFICANCE These results suggested that TP receptor signaling facilitates liver repair and sinusoidal restoration from acute liver injury through HGF release from platelets adhering to the sinusoids.
Collapse
Affiliation(s)
- Tsutomu Minamino
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan; Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Yoshiya Ito
- Department of Surgery, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Hirotoki Ohkubo
- Department of Surgery, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Yuki Shimuzu
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan; Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Ken Kojo
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan; Department of Surgery, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Nobuyuki Nishizwa
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan; Department of Surgery, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Shuh Narumiya
- Innovation Center for Immunoregulation Technologies and Drugs (AK project), Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Wasaburo Koizumi
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine, Kanagawa 252-0374, Japan.
| |
Collapse
|
152
|
Usta OB, McCarty WJ, Bale S, Hegde M, Jindal R, Bhushan A, Golberg I, Yarmush ML. Microengineered cell and tissue systems for drug screening and toxicology applications: Evolution of in-vitro liver technologies. TECHNOLOGY 2015; 3:1-26. [PMID: 26167518 PMCID: PMC4494128 DOI: 10.1142/s2339547815300012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The liver performs many key functions, the most prominent of which is serving as the metabolic hub of the body. For this reason, the liver is the focal point of many investigations aimed at understanding an organism's toxicological response to endogenous and exogenous challenges. Because so many drug failures have involved direct liver toxicity or other organ toxicity from liver generated metabolites, the pharmaceutical industry has constantly sought superior, predictive in-vitro models that can more quickly and efficiently identify problematic drug candidates before they incur major development costs, and certainly before they are released to the public. In this broad review, we present a survey and critical comparison of in-vitro liver technologies along a broad spectrum, but focus on the current renewed push to develop "organs-on-a-chip". One prominent set of conclusions from this review is that while a large body of recent work has steered the field towards an ever more comprehensive understanding of what is needed, the field remains in great need of several key advances, including establishment of standard characterization methods, enhanced technologies that mimic the in-vivo cellular environment, and better computational approaches to bridge the gap between the in-vitro and in-vivo results.
Collapse
Affiliation(s)
- O B Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - W J McCarty
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - S Bale
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M Hegde
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - R Jindal
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - A Bhushan
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - I Golberg
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| | - M L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA ; Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
153
|
Rozance PJ, Anderson M, Martinez M, Fahy A, Macko AR, Kailey J, Seedorf GJ, Abman SH, Hay WW, Limesand SW. Placental insufficiency decreases pancreatic vascularity and disrupts hepatocyte growth factor signaling in the pancreatic islet endothelial cell in fetal sheep. Diabetes 2015; 64:555-64. [PMID: 25249573 PMCID: PMC4303968 DOI: 10.2337/db14-0462] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hepatocyte growth factor (HGF) and vascular endothelial growth factor A (VEGFA) are paracrine hormones that mediate communication between pancreatic islet endothelial cells (ECs) and β-cells. Our objective was to determine the impact of intrauterine growth restriction (IUGR) on pancreatic vascularity and paracrine signaling between the EC and β-cell. Vessel density was less in IUGR pancreata than in controls. HGF concentrations were also lower in islet EC-conditioned media (ECCM) from IUGR, and islets incubated with control islet ECCM responded by increasing insulin content, which was absent with IUGR ECCM. The effect of ECCM on islet insulin content was blocked with an inhibitory anti-HGF antibody. The HGF receptor was not different between control and IUGR islets, but VEGFA was lower and the high-affinity VEGF receptor was higher in IUGR islets and ECs, respectively. These findings show that paracrine actions from ECs increase islet insulin content, and in IUGR ECs, secretion of HGF was diminished. Given the potential feed-forward regulation of β-cell VEGFA and islet EC HGF, these two growth factors are highly integrated in normal pancreatic islet development, and this regulation is decreased in IUGR fetuses, resulting in lower pancreatic islet insulin concentrations and insulin secretion.
Collapse
Affiliation(s)
- Paul J Rozance
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO Perinatal Research Center, University of Colorado School of Medicine, Aurora, CO
| | - Miranda Anderson
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Marina Martinez
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Anna Fahy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Antoni R Macko
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| | - Jenai Kailey
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO Perinatal Research Center, University of Colorado School of Medicine, Aurora, CO
| | - Gregory J Seedorf
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO
| | - Steven H Abman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO
| | - William W Hay
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ
| |
Collapse
|
154
|
Zhao X, Zhao Q, Luo Z, Yu Y, Xiao N, Sun X, Cheng L. Spontaneous immortalization of mouse liver sinusoidal endothelial cells. Int J Mol Med 2015; 35:617-24. [PMID: 25585915 PMCID: PMC4314414 DOI: 10.3892/ijmm.2015.2067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 12/30/2014] [Indexed: 12/27/2022] Open
Abstract
The spontaneous immortalization of cells in vitro is a rare event requiring genomic instability, such as alterations in chromosomes and mutations in genes. In the present study, we report a spontaneously immortalized liver sinusoidal endothelial cell (LSEC) line generated from mouse liver. These immortalized LSECs showed typical LSEC characteristics with the structure of transcellular fenestrations, the expression of von Willebrand factor (VWF) and the ability to uptake DiI-acetylated-low density lipoprotein (DiI-Ac-LDL). However, these immortalized LSECs lost the ability to form capillary-like structures, and showed clonal and multilayer growth without contact inhibition. Moreover, their proliferation rate increased with the increase in the number of passages. In addition, these cells obained the expression of CD31 and desmin, and showed an upregulation of p53 protein expression; however, their karyotype was normal, and they could not form colonies in soft agar or tumors in SCID mice. In conclusion, in the present study, we successfully established a spontaneously immortalized LSEC line.
Collapse
Affiliation(s)
- Xiuhua Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shanxi 710004, P.R. China
| | - Qian Zhao
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhen Luo
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yan Yu
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Na Xiao
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xuan Sun
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Lamei Cheng
- Department of Adult Stem Cells, Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
155
|
Abstract
In this issue of Cancer Discovery, Horwitz and colleagues identified a subtype of hepatocelluar carcinoma (HCC) bearing VEGFA genomic amplification that is particularly sensitive to VEGFA inhibition and is also more sensitive to sorafenib treatment. Taken conjointly, these data suggest that VEGFA genomic amplification can be used as a biomarker for personalized treatment of HCC with sorafenib.
Collapse
Affiliation(s)
- Xiaolin Luo
- Authors' Affiliation: Department of Pathology and Division of Biological Sciences, University of California San Diego, La Jolla, California
| | - Gen-Sheng Feng
- Authors' Affiliation: Department of Pathology and Division of Biological Sciences, University of California San Diego, La Jolla, California
| |
Collapse
|
156
|
Gao R, Wu W, Xiang J, Lv Y, Zheng X, Chen Q, Wang H, Wang B, Liu Z, Ma F. Hepatocyte culture in autologous decellularized spleen matrix. Organogenesis 2015; 11:16-29. [PMID: 25664568 PMCID: PMC4594376 DOI: 10.1080/15476278.2015.1011908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/10/2014] [Accepted: 01/18/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND AIMS Using decellularized scaffold to reengineer liver tissue is a promising alternative therapy for end-stage liver diseases. Though the decellularized human liver matrix is the ideal scaffold for reconstruction of the liver theoretically, the shortage of liver donors is still an obstacle for potential clinical application. Therefore, an appropriate alternative scaffold is needed. In the present study, we used a tissue engineering approach to prepare a rat decellularized spleen matrix (DSM) and evaluate the effectiveness of this DSM for primary rat hepatocytes culture. METHODS Rat decellularized spleen matrix (DSM) was prepared by perfusion of a series of detergents through spleen vasculature. DSM was characterized by residual DNA and specific extracellular matrix distribution. Thereafter, primary rat hepatocytes were cultured in the DSM in a 3-dimensional dynamic culture system, and liver cell survival and biological functions were evaluated by comparison with 3-dimensional sandwich culture and also with cultured in decellularized liver matrix (DLM). RESULTS Our research found that DSM did not exhibit any cellular components, but preserved the main extracellular matrix and the intact vasculature evaluated by DNA detection, histology, immunohistochemical staining, vessel corrosion cast and upright metallurgical microscope. Moreover, the method of DSM preparation procedure was relatively simple with high success rate (100%). After seeding primary hepatocytes in DSM, the cultured hepatocytes survived inside DSM with albumin synthesis and urea secretion within 10 d. Additionally, hepatocytes in dynamic culture medium had better biological functions at day 10 than that in sandwich culture. Albumin synthesis was 85.67 ± 6.34 μg/10(7) cell/24h in dynamic culture in DSM compared to 62.43 ± 4.59 μg/10(7) cell/24h in sandwich culture (P < 0.01) and to 87.54 ± 5.25 μg/10(7) cell/24h in DLM culture (P > 0.05); urea release was 32.14 ± 8.62 μg/10(7) cell/24h in dynamic culture in DSM compared to 20.47 ± 4.98 μg/10(7) cell/24h in sandwich culture (P < 0.05) and to 37.38 ± 7.29 μg/10(7) cell/24h cultured in DLM (P > 0.05). CONCLUSION The present study demonstrates that DSM can be prepared successfully using a tissue engineering approach. The DSM is an appropriate scaffold for primary hepatocytes culture.
Collapse
Affiliation(s)
- Rui Gao
- Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Wanquan Wu
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Junxi Xiang
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Yi Lv
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Xinglong Zheng
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Qian Chen
- Department of Bio-Medical Sciences; Philadelphia College of Osteopathic Medicine; Philadelphia, PA USA
| | - Haohua Wang
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Bo Wang
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Zhengwen Liu
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Infectious Diseases; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| | - Feng Ma
- Institute of Advanced Surgical Techniques and Tissue Engineering Research; Xi'an Jiaotong University; Xi'an, Shaanxi, China
- Department of Hepatobiliary Surgery; First Hospital of Medical College; Xi'an Jiaotong University; Xi'an, Shaanxi, China
| |
Collapse
|
157
|
Horwitz E, Stein I, Ben-Neriah Y, Pikarsky E. Animal model studies indicate a candidate biomarker for sorafenib treatment of hepatocellular carcinoma. Mol Cell Oncol 2015; 2:e968028. [PMID: 27308384 DOI: 10.4161/23723548.2014.968028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 09/17/2014] [Accepted: 08/18/2014] [Indexed: 11/19/2022]
Abstract
In contrast to common genomic amplifications that support cancer cell growth by rewiring intracellular signaling, VEGFA amplification drives tumor cell proliferation via the tumor microenvironment. VEGFA amplification is present in a subset of mouse and human hepatocellular carcinomas (HCCs) that appear to be particularly sensitive to sorafenib treatment, indicating its potential value as a biomarker for HCC treatment.
Collapse
Affiliation(s)
- Elad Horwitz
- The Lautenberg Center for Immunology; IMRIC, Hebrew University - Hadassah Medical School ; Jerusalem, Israel
| | - Ilan Stein
- The Lautenberg Center for Immunology; IMRIC, Hebrew University - Hadassah Medical School; Jerusalem, Israel; Department of Pathology; Hadassah-Hebrew University Medical Center; Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology; IMRIC, Hebrew University - Hadassah Medical School ; Jerusalem, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology; IMRIC, Hebrew University - Hadassah Medical School; Jerusalem, Israel; Department of Pathology; Hadassah-Hebrew University Medical Center; Jerusalem, Israel
| |
Collapse
|
158
|
Muto J, Shirabe K, Sugimachi K, Maehara Y. Review of angiogenesis in hepatocellular carcinoma. Hepatol Res 2015; 45:1-9. [PMID: 24533487 DOI: 10.1111/hepr.12310] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/27/2014] [Accepted: 02/03/2014] [Indexed: 12/27/2022]
Abstract
Hepatocellular carcinoma (HCC) is a hypervascular tumor, and its vascularity is unique and greatly different from peripheral parenchyma of liver. Afferent and efferent vessels of HCC lesions come to differ as the lesion develops. The characteristic of the flow regulates the common style of metastasis. The portal tract of the HCC lesion is the first site of the intrahepatic metastasis, because cancer cells roll into the portal vein via efferent flow. On microscopic observation, HCC displays marked vascular abnormalities, arteriogenesis and capillarization. Arteriogenesis is defined as the growth of functional collateral arteries covered with smooth muscle cells from pre-existing arteries. Sinusoidal capillarization involves the transformation of fenestrated hepatic sinusoids into continuous capillaries. Several angiogenic factors have been reported, and some of them are studied as prognostic factors or target molecules of chemotherapeutic reagents. However, the mechanism of neovascularization during HCC development is still unclear. This review discusses the characteristics of angiogenesis in HCC and known angiogenic factors of HCC.
Collapse
Affiliation(s)
- Jun Muto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | |
Collapse
|
159
|
Abstract
The liver is the largest internal organ in mammals, serving a wide spectrum of vital functions. Loss of liver function due to drug toxicity, progressive fatty liver disease, or viral infection is a major cause of death in the United States of America. Pharmaceutical and cosmetic toxicity screening, basic research and the development of bioartificial liver devices require long-term hepatocyte culture techniques that sustain hepatocyte morphology and function. In recent years, several techniques have been developed that can support high levels of liver-specific gene expression, metabolic function, and synthetic activity for several weeks in culture. These include the collagen double gel configuration, hepatocyte spheroids, coculture with nonparenchymal cells, and micropatterned cocultures. This chapter will cover the current status of hepatocyte culture techniques, including media formulation, oxygen supply, and heterotypic cell-cell interactions.
Collapse
Affiliation(s)
- Merav Cohen
- Department of Cell and Developmental Biology, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | |
Collapse
|
160
|
Ramasamy SK, Kusumbe AP, Adams RH. Regulation of tissue morphogenesis by endothelial cell-derived signals. Trends Cell Biol 2014; 25:148-57. [PMID: 25529933 DOI: 10.1016/j.tcb.2014.11.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) form an extensive network of blood vessels that has numerous essential functions in the vertebrate body. In addition to their well-established role as a versatile transport network, blood vessels can induce organ formation or direct growth and differentiation processes by providing signals in a paracrine (angiocrine) fashion. Tissue repair also requires the local restoration of vasculature. ECs are emerging as important signaling centers that coordinate regeneration and help to prevent deregulated, disease-promoting processes. Vascular cells are also part of stem cell niches and have key roles in hematopoiesis, bone formation, and neurogenesis. Here, we review these newly identified roles of ECs in the regulation of organ morphogenesis, maintenance, and regeneration.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, University of Münster, Faculty of Medicine, D-48149 Münster, Germany
| | - Anjali P Kusumbe
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, University of Münster, Faculty of Medicine, D-48149 Münster, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, University of Münster, Faculty of Medicine, D-48149 Münster, Germany.
| |
Collapse
|
161
|
Increased VEGFR2 and MMP9 protein levels are associated with epithelial dysplasia grading. Pathol Res Pract 2014; 210:959-64. [DOI: 10.1016/j.prp.2014.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/17/2013] [Accepted: 06/24/2014] [Indexed: 01/10/2023]
|
162
|
Géraud C, Koch PS, Damm F, Schledzewski K, Goerdt S. Der metastatische Zyklus: metastatische Nischen und Tumorzell-Dissemination. J Dtsch Dermatol Ges 2014. [DOI: 10.1111/ddg.12451_suppl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Cyrill Géraud
- Klinik für Dermatologie; Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät der Universität Heidelberg; Mannheim
| | - Philipp S Koch
- Klinik für Dermatologie; Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät der Universität Heidelberg; Mannheim
| | - Friederike Damm
- Klinik für Dermatologie; Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät der Universität Heidelberg; Mannheim
| | - Kai Schledzewski
- Klinik für Dermatologie; Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät der Universität Heidelberg; Mannheim
| | - Sergij Goerdt
- Klinik für Dermatologie; Venerologie und Allergologie, Universitätsklinikum Mannheim, Medizinische Fakultät der Universität Heidelberg; Mannheim
| |
Collapse
|
163
|
Abstract
Liver regeneration after partial hepatectomy is the only example of a regenerative process in mammals in which the organ/body weight ratio returns to 100% of the original when the process is complete. The adjustment of liver weight to the needs of the body suggests a complicated set of control points, a 'hepatostat'. There has been much progress in elucidation of mechanisms involved in initiation of liver regeneration. More recent studies have focused on termination pathways, because these may be the underlying controls of the hepatostat and their elimination may be relevant to hepatic neoplasia. When the standard regenerative process is thwarted due to failure of either hepatocytes or biliary epithelial cells to proliferate, each of the two epithelial compartments can function as a source of facultative stem cells for the other.
Collapse
Affiliation(s)
- George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Bioscience Tower South, Pittsburgh, PA 15261, USA
| |
Collapse
|
164
|
Géraud C, Koch PS, Damm F, Schledzewski K, Goerdt S. The metastatic cycle: metastatic niches and cancer cell dissemination. J Dtsch Dermatol Ges 2014; 12:1012-9. [PMID: 25307562 DOI: 10.1111/ddg.12451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/19/2014] [Indexed: 12/23/2022]
Abstract
The concept of a unidirectional cascade of metastatic events has been replaced in recent years by the metastatic cycle - the concept of a dynamic feed forward cycle of metastatic niches that evolve upon reciprocal interactions with the primary tumor and disseminating cancer cells. Primary tumors interact with pre-metastatic sites preparing organ-specific pre-metastatic niches. Metastasis-initiating cells home to and succumb to interactions with developing organ-specific metastatic niches, and secondary recirculating cancer cells interact back with the primary. Metastatic tropism as well as metastatic disease progression are a result of this feed forward cycle of dynamic, reciprocal interactions of cancer cells with their diverse metastatic niches. A better understanding of the multifaceted contributions of the organ-specific metastatic niches and their complex changes on cancer cell dissemination and of the mutual effects of the cellular and molecular mechanisms involved will open new avenues to better therapies for hitherto intractable progressive disease states of cancer patients and for adjuvant treatment options preventing relapses in tumor-free patients.
Collapse
Affiliation(s)
- Cyrill Géraud
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | |
Collapse
|
165
|
Vascular endothelial growth factor: therapeutic possibilities and challenges for the treatment of ischemia. Cytokine 2014; 71:385-93. [PMID: 25240960 DOI: 10.1016/j.cyto.2014.08.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/02/2014] [Accepted: 08/19/2014] [Indexed: 12/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a notable chemokine that plays critical roles in angiogenesis and vasculogenesis. The contemporary body of literature contains a substantial amount of information regarding its chemical properties as well as its fundamental role in vascular development. Studies strongly indicate its potential use as a therapeutic agent, especially in the vascular restoration of injured and ischemic tissues. VEGF therapy could be most beneficial for diseases whose pathologies revolve around tissue inflammation and necrosis, such as myocardial infarction and stroke, as well as ischemic bowel diseases such as acute mesenteric ischemia and necrotizing enterocolitis. However, a delicate balance exists between the therapeutic benefits of VEGF and the hazards of tumor growth and neo-angiogenesis. Effective future research surrounding VEGF may allow for the development of effective therapies for ischemia which simultaneously limit its more deleterious side effects. This review will: (1) summarize the current understanding of the molecular aspects and function of VEGF, (2) review potential benefits of its use in medical therapy, (3) denote its role in tumorigenesis and inflammation when overexpressed, and (4) elucidate the qualities which make it a viable compound of study for diagnostic and therapeutic applications.
Collapse
|
166
|
Lee YJ, Park HJ, Woo SY, Park EM, Kang JL. RhoA/phosphatidylinositol 3-kinase/protein kinase B/mitogen-activated protein kinase signaling after growth arrest-specific protein 6/mer receptor tyrosine kinase engagement promotes epithelial cell growth and wound repair via upregulation of hepatocyte growth factor in macrophages. J Pharmacol Exp Ther 2014; 350:563-77. [PMID: 24939420 DOI: 10.1124/jpet.114.215673] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Growth arrest-specific protein 6 (Gas6)/Mer receptor tyrosine kinase (Mer) signaling modulates cytokine secretion and helps to regulate the immune response and apoptotic cell clearance. Signaling pathways that activate an epithelial growth program in macrophages are still poorly defined. We report that Gas6/Mer/RhoA signaling can induce the production of epithelial growth factor hepatic growth factor (HGF) in macrophages, which ultimately promotes epithelial cell proliferation and wound repair. The RhoA/protein kinase B (Akt)/mitogen-activated protein (MAP) kinases, including p38 MAP kinase, extracellular signal-regulated protein kinase, and Jun NH2-terminal kinase axis in RAW 264.7 cells, was identified as Gas6/Mer downstream signaling pathway for the upregulation of HGF mRNA and protein. Conditioned medium from RAW 264.7 cells that had been exposed to Gas6 or apoptotic cells enhanced epithelial cell proliferation of the epithelial cell line LA-4 and wound closure. Cotreatment with an HGF receptor-blocking antibody or c-Met antagonist downregulated this enhancement. Inhibition of Mer with small interfering RNA (siRNA) or the RhoA/Rho kinase pathway by RhoA siRNA or Rho kinase pharmacologic inhibitor suppressed Gas6-induced HGF mRNA and protein expression in macrophages and blocked epithelial cell proliferation and wound closure induced by the conditioned medium. Our data provide evidence that macrophages can be reprogrammed by Gas6 to promote epithelial proliferation and wound repair via HGF, which is induced by the Mer/RhoA/Akt/MAP kinase pathway. Thus, defects in Gas6/Mer/RhoA signaling in macrophages may delay tissue repair after injury to the alveolar epithelium.
Collapse
Affiliation(s)
- Ye-Ji Lee
- Department of Physiology (Y.-J.L., H.-J.P, S.-Y.W., J.L.K.), Department of Pharmacology (E.-M.P.), Department of Microbiology A (S.-Y.W.), and Tissue Injury Defense Research Center (H.-J.P., S.-Y.W., E.-M.P., J.L.K.), Global Top5 Research Program (J.L.K.), School of Medicine, Ewha Womans University, Seoul, Korea
| | - Hyun-Jung Park
- Department of Physiology (Y.-J.L., H.-J.P, S.-Y.W., J.L.K.), Department of Pharmacology (E.-M.P.), Department of Microbiology A (S.-Y.W.), and Tissue Injury Defense Research Center (H.-J.P., S.-Y.W., E.-M.P., J.L.K.), Global Top5 Research Program (J.L.K.), School of Medicine, Ewha Womans University, Seoul, Korea
| | - So-Youn Woo
- Department of Physiology (Y.-J.L., H.-J.P, S.-Y.W., J.L.K.), Department of Pharmacology (E.-M.P.), Department of Microbiology A (S.-Y.W.), and Tissue Injury Defense Research Center (H.-J.P., S.-Y.W., E.-M.P., J.L.K.), Global Top5 Research Program (J.L.K.), School of Medicine, Ewha Womans University, Seoul, Korea
| | - Eun-Mi Park
- Department of Physiology (Y.-J.L., H.-J.P, S.-Y.W., J.L.K.), Department of Pharmacology (E.-M.P.), Department of Microbiology A (S.-Y.W.), and Tissue Injury Defense Research Center (H.-J.P., S.-Y.W., E.-M.P., J.L.K.), Global Top5 Research Program (J.L.K.), School of Medicine, Ewha Womans University, Seoul, Korea
| | - Jihee Lee Kang
- Department of Physiology (Y.-J.L., H.-J.P, S.-Y.W., J.L.K.), Department of Pharmacology (E.-M.P.), Department of Microbiology A (S.-Y.W.), and Tissue Injury Defense Research Center (H.-J.P., S.-Y.W., E.-M.P., J.L.K.), Global Top5 Research Program (J.L.K.), School of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
167
|
Al-Maghrabi J, Gomaa W, Buhmeida A, Qari Y, Al-Qahtani M, Al-Ahwal M. Prognostic significance of VEGFR1/Flt-1 immunoexpression in colorectal carcinoma. Tumour Biol 2014; 35:9045-9051. [PMID: 24908415 DOI: 10.1007/s13277-014-2124-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/20/2014] [Indexed: 01/13/2023] Open
Abstract
Colorectal carcinoma (CRC) is a major cause of morbidity and mortality. Vascular endothelial growth factor 1/Fms-like tyrosine kinase 1 (VEGFR1/Flt-1) regulates monocyte migration, recruits endothelial cell progenitors, increases the adhesive properties of natural killer cells and induces of growth factors. Flt-1 is expressed on tumour cells and has been implicated in tumour growth and progression. The objective of this study is to address the relation of Flt-1 expression to tumour prognostication. Paraffin blocks from 143 primary CRC and 48 regional nodal metastases were retrieved from the archives of the Department of Pathology at King Abdulaziz University. Tissue microarrays were designed and constructed. Immunohistochemistry for Flt-1 was performed. Staining intensity and extent of staining were assessed and combined. Results were dichotomised as low expression and high expression. Flt-1 was overexpressed in primary tumours and nodal metastasis (p < 0.001 and 0.001) with no difference between primary and nodal metastasis (p = 0.690). Flt-1 immunoexpression was not associated with the clinicopathological parameters. Flt-1 overexpression was an independent predictor of positive margin status, positive lymphovascular invasion and local disease recurrence (p < 0.001, p < 0.001 and p = 0.003, respectively). Flt-1 was not associated with survival (log-rank = 0.003, p = 0.959). Flt-1 was overexpressed in primary CRC and their nodal metastases. Flt-1 expression was an independent predictor of margin status, lymphovascular invasion and local disease recurrence. Therefore, expression profiling of Flt-1 seems to have a prognostic potential in CRC. However, to elucidate the association of overexpression of Flt-1 with tumour characteristics and prognostication, more in vivo and in vitro molecular investigations are recommended.
Collapse
Affiliation(s)
- Jaudah Al-Maghrabi
- Scientific Chair for Colorectal Cancer, King Abdulaziz University, Jeddah, Saudi Arabia,
| | | | | | | | | | | |
Collapse
|
168
|
Ohkubo H, Ito Y, Minamino T, Eshima K, Kojo K, Okizaki SI, Hirata M, Shibuya M, Watanabe M, Majima M. VEGFR1-positive macrophages facilitate liver repair and sinusoidal reconstruction after hepatic ischemia/reperfusion injury. PLoS One 2014; 9:e105533. [PMID: 25162491 PMCID: PMC4146544 DOI: 10.1371/journal.pone.0105533] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 07/22/2014] [Indexed: 01/19/2023] Open
Abstract
Liver repair after acute liver injury is characterized by hepatocyte proliferation, removal of necrotic tissue, and restoration of hepatocellular and hepatic microvascular architecture. Macrophage recruitment is essential for liver tissue repair and recovery from injury; however, the underlying mechanisms are unclear. Signaling through vascular endothelial growth factor receptor 1 (VEGFR1) is suggested to play a role in macrophage migration and angiogenesis. The aim of the present study was to examine the role of VEGFR1 in liver repair and sinusoidal reconstruction after hepatic ischemia/reperfusion (I/R). VEGFR1 tyrosine kinase knockout mice (VEGFR1 TK-/- mice) and wild-type (WT) mice were subjected to hepatic warm I/R, and the processes of liver repair and sinusoidal reconstruction were examined. Compared with WT mice, VEGFR1 TK-/- mice exhibited delayed liver repair after hepatic I/R. VEGFR1-expressing macrophages recruited to the injured liver showed reduced expression of epidermal growth factor (EGF). VEGFR1 TK-/- mice also showed evidence of sustained sinusoidal functional and structural damage, and reduced expression of pro-angiogenic factors. Treatment of VEGFR1 TK-/- mice with EGF attenuated hepatoceullar and sinusoidal injury during hepatic I/R. VEGFR1 TK-/- bone marrow (BM) chimeric mice showed impaired liver repair and sinusoidal reconstruction, and reduced recruitment of VEGFR1-expressing macrophages to the injured liver. VEGFR1-macrophages recruited to the liver during hepatic I/R contribute to liver repair and sinusoidal reconstruction. VEGFR1 activation is a potential therapeutic strategy for promoting liver repair and sinusoidal restoration after acute liver injury.
Collapse
Affiliation(s)
- Hirotoki Ohkubo
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Yoshiya Ito
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tsutomu Minamino
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Koji Eshima
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Ken Kojo
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shin-ichiro Okizaki
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Mitsuhiro Hirata
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masabumi Shibuya
- Gakubunkan Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Masahiko Watanabe
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
169
|
Vainshtein JM, Kabarriti R, Mehta KJ, Roy-Chowdhury J, Guha C. Bone marrow-derived stromal cell therapy in cirrhosis: clinical evidence, cellular mechanisms, and implications for the treatment of hepatocellular carcinoma. Int J Radiat Oncol Biol Phys 2014; 89:786-803. [PMID: 24969793 DOI: 10.1016/j.ijrobp.2014.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 01/18/2023]
Abstract
Current treatment options for hepatocellular carcinoma (HCC) are often limited by the presence of underlying liver disease. In patients with liver cirrhosis, surgery, chemotherapy, and radiation therapy all carry a high risk of hepatic complications, ranging from ascites to fulminant liver failure. For patients receiving radiation therapy, cirrhosis dramatically reduces the already limited radiation tolerance of the liver and represents the most important clinical risk factor for the development of radiation-induced liver disease. Although improvements in conformal radiation delivery techniques have improved our ability to safely irradiate confined areas of the liver to increasingly higher doses with excellent local disease control, patients with moderate-to-severe liver cirrhosis continue to face a shortage of treatment options for HCC. In recent years, evidence has emerged supporting the use of bone marrow-derived stromal cells (BMSCs) as a promising treatment for liver cirrhosis, with several clinical studies demonstrating sustained improvement in clinical parameters of liver function after autologous BMSC infusion. Three predominant populations of BMSCs, namely hematopoietic stem cells, mesenchymal stem cells, and endothelial progenitor cells, seem to have therapeutic potential in liver injury and cirrhosis. Preclinical studies of BMSC transplantation have identified a range of mechanisms through which these cells mediate their therapeutic effects, including hepatocyte transdifferentiation and fusion, paracrine stimulation of hepatocyte proliferation, inhibition of activated hepatic stellate cells, enhancement of fibrolytic matrix metalloproteinase activity, and neovascularization of regenerating liver. By bolstering liver function in patients with underlying Child's B or C cirrhosis, autologous BMSC infusion holds great promise as a therapy to improve the safety, efficacy, and utility of surgery, chemotherapy, and hepatic radiation therapy in the treatment of HCC.
Collapse
Affiliation(s)
| | - Rafi Kabarriti
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Keyur J Mehta
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jayanta Roy-Chowdhury
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Genetics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
170
|
Sohn BS, Park SJ, Kim JE, Kim KP, Hong YS, Suh C, Kim YS, Kim SY, Im SA, Kim SY, Kim JH, Ahn JB, Park YS, Kim TW. Single-nucleotide polymorphisms in the vascular endothelial growth factor pathway and outcomes of patients treated with first-line cytotoxic chemotherapy combined with bevacizumab for advanced colorectal cancer. Oncology 2014; 87:280-92. [PMID: 25139485 DOI: 10.1159/000365593] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/30/2014] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate the association between the efficacy of first-line cytotoxic chemotherapy plus bevacizumab and single-nucleotide polymorphisms (SNPs) of angiogenic genes in patients with advanced colorectal cancer (CRC). METHODS DNA was extracted from blood samples of 125 patients, and 12 SNPs were evaluated for association with the objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). RESULTS The vascular endothelial growth factor A (VEGFA) rs833061 T/T was associated with superior ORR compared to its alternative genotypes (75.9 vs. 50.8%; p = 0.008), and the interleukin 8 rs4073 A/A genotype tended to be associated with poor ORR (45.0 vs. 66.0%; p = 0.067). The median PFS and OS were superior in patients with the fms-related tyrosine kinase 1 (FLT1) rs9513070 A/A genotype (8.7 vs. 6.6 months; p = 0.001 and 26.4 vs. 16.1 months; p = 0.038, respectively). The kinase insert domain receptor rs1531289 G/G genotype tended to be associated with improved PFS (8.0 vs. 7.1 months; p = 0.069). In haplotype analysis, the FLT1 rs9513070/rs9554320/rs9582036 GCA haplotype was associated with inferior PFS and OS (p = 0.004 and p = 0.041, respectively). CONCLUSION The VEGFA rs833061 SNP is associated with the ORR, and the FLT1 rs9513070 SNP and FLT1 GCA haplotypes are associated with PFS and OS in advanced CRC patients treated with cytotoxic chemotherapy plus bevacizumab.
Collapse
Affiliation(s)
- Byeong Seok Sohn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Caralt M, Velasco E, Lanas A, Baptista PM. Liver bioengineering: from the stage of liver decellularized matrix to the multiple cellular actors and bioreactor special effects. Organogenesis 2014; 10:250-9. [PMID: 25102189 DOI: 10.4161/org.29892] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Liver bioengineering has been a field of intense research and popular excitement in the past decades. It experiences great interest since the introduction of whole liver acellular scaffolds generated by perfusion decellularization (1-3). Nevertheless, the different strategies developed so far have failed to generate hepatic tissue in vitro bioequivalent to native liver tissue. Even notable novel strategies that rely on iPSC-derived liver progenitor cells potential to self-organize in association with endothelial cells in hepatic organoids are lacking critical components of the native tissue (e.g., bile ducts, functional vascular network, hepatic microarchitecture, etc) (4). Hence, it is vital to understand the strengths and short comes of our current strategies in this quest to re-create liver organogenesis in vitro. To shed some light into these issues, this review describes the different actors that play crucial roles in liver organogenesis and highlights the steps still missing to successfully generate whole livers and hepatic organoids in vitro for multiple applications.
Collapse
Affiliation(s)
- Mireia Caralt
- Vall d'Hebron University Hospital; Universitat Autònoma de Barcelona; Barcelona, Spain
| | | | - Angel Lanas
- University of Zaragoza; Zaragoza, Spain; IIS Aragón; CIBERehd; Zaragoza, Spain; Aragon Health Sciences Institute (IACS); Zaragoza, Spain
| | - Pedro M Baptista
- University of Zaragoza; Zaragoza, Spain; IIS Aragón; CIBERehd; Zaragoza, Spain; Aragon Health Sciences Institute (IACS); Zaragoza, Spain
| |
Collapse
|
172
|
Preventive effects of dexmedetomidine on the liver in a rat model of acid-induced acute lung injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:621827. [PMID: 25165710 PMCID: PMC4138784 DOI: 10.1155/2014/621827] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 07/08/2014] [Accepted: 07/17/2014] [Indexed: 01/20/2023]
Abstract
The aim of this study was to examine whether dexmedetomidine improves acute liver injury in a rat model. Twenty-eight male Wistar albino rats weighing 300–350 g were allocated randomly to four groups. In group 1, normal saline (NS) was injected into the lungs and rats were allowed to breathe spontaneously. In group 2, rats received standard ventilation (SV) in addition to NS. In group 3, hydrochloric acid was injected into the lungs and rats received SV. In group 4, rats received SV and 100 µg/kg intraperitoneal dexmedetomidine before intratracheal HCl instillation. Blood samples and liver tissue specimens were examined by biochemical, histopathological, and immunohistochemical methods. Acute lung injury (ALI) was found to be associated with increased malondialdehyde (MDA), total oxidant activity (TOA), oxidative stress index (OSI), and decreased total antioxidant capacity (TAC). Significantly decreased MDA, TOA, and OSI levels and significantly increased TAC levels were found with dexmedetomidine injection in group 4 (P < 0.05). The highest histologic injury scores were detected in group 3. Enhanced hepatic vascular endothelial growth factor (VEGF) expression and reduced CD68 expression were found in dexmedetomidine group compared with the group 3. In conclusion, the presented data provide the first evidence that dexmedetomidine has a protective effect on experimental liver injury induced by ALI.
Collapse
|
173
|
Huebert RC, Shah VH. Sinusoidal endothelial cells direct traffic at the intersection of regeneration and fibrosis. Hepatology 2014; 60:754-6. [PMID: 24615996 PMCID: PMC4110160 DOI: 10.1002/hep.27116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/04/2014] [Accepted: 03/04/2014] [Indexed: 12/29/2022]
Abstract
Chemical or traumatic damage to the liver is frequently associated with aberrant healing (fibrosis) that overrides liver regeneration. The mechanism by which hepatic niche cells differentially modulate regeneration and fibrosis during liver repair remains to be defined. Hepatic vascular niche predominantly represented by liver sinusoidal endothelial cells deploys paracrine trophogens, known as angiocrine factors, to stimulate regeneration. Nevertheless, it is not known how pro-regenerative angiocrine signals from liver sinusoidal endothelial cells is subverted to promote fibrosis. Here, by combining an inducible endothelial-cell-specific mouse gene deletion strategy and complementary models of acute and chronic liver injury, we show that divergent angiocrine signals from liver sinusoidal endothelial cells stimulate regeneration after immediate injury and provoke fibrosis after chronic insult. The pro-fibrotic transition of vascular niche results from differential expression of stromal-derived factor-1 receptors, CXCR7 and CXCR4 in liver sinusoidal endothelial cells. After acute injury, CXCR7 upregulation in liver sinusoidal endothelial cells acts with CXCR4 to induce transcription factor Id1, deploying pro-regenerative angiocrine factors and triggering regeneration. Inducible deletion of Cxcr7 in sinusoidal endothelial cells (Cxcr7iΔEC/iΔEC) from the adult mouse liver impaired liver regeneration by diminishing Id1-mediated production of angiocrine factors. By contrast, after chronic injury inflicted by iterative hepatotoxin (carbon tetrachloride) injection and bile duct ligation, constitutive FGFR1 signaling in liver sinusoidal endothelial cells counterbalanced CXCR7-dependent pro-regenerative response and augmented CXCR4 expression. This predominance of CXCR4 over CXCR7 expression shifted angiocrine response of liver sinusoidal endothelial cells, stimulating proliferation of desmin+ hepatic stellate-like cells and enforcing a pro-fibrotic vascular niche. Endothelial-cell-specific ablation of either Fgfr1 (Fgfr1iΔEC/iΔEC) or Cxcr4 (Cxcr4iΔEC/iΔEC) in mice restored the pro-regenerative pathway and prevented FGFR1-mediated maladaptive subversion of angiocrine factors. Similarly, selective CXCR7 activation in liver sinusoidal endothelial cells abrogated fibrogenesis. Thus, we demonstrate that in response to liver injury, differential recruitment of pro-regenerative CXCR7-Id1 versus pro-fibrotic FGFR1-CXCR4 angiocrine pathways in vascular niche balances regeneration and fibrosis. These results provide a therapeutic roadmap to achieve hepatic regeneration without provoking fibrosis.
Collapse
Affiliation(s)
- Robert C. Huebert
- Gastroenterology Research Unit; Mayo Clinic and Foundation; Rochester MN
| | - Vijay H. Shah
- Gastroenterology Research Unit; Mayo Clinic and Foundation; Rochester MN
| |
Collapse
|
174
|
Stimulatory effect of vascular endothelial growth factor on progesterone production and survivability of cultured bubaline luteal cells. Anim Reprod Sci 2014; 148:251-9. [DOI: 10.1016/j.anireprosci.2014.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/14/2014] [Accepted: 06/19/2014] [Indexed: 11/17/2022]
|
175
|
Géraud C, Koch PS, Goerdt S. Vaskuläre Nischen: Endothelzellen als multifunktionale gewebe- und standortspezifische Teamplayer im gesunden und erkrankten Organismus. J Dtsch Dermatol Ges 2014. [DOI: 10.1111/ddg.12402_suppl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Cyrill Géraud
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University; Mannheim Germany
| | - Philipp-S. Koch
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University; Mannheim Germany
| | - Sergij Goerdt
- Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University; Mannheim Germany
| |
Collapse
|
176
|
Yagai T, Miyajima A, Tanaka M. Semaphorin 3E secreted by damaged hepatocytes regulates the sinusoidal regeneration and liver fibrosis during liver regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2250-9. [PMID: 24930441 DOI: 10.1016/j.ajpath.2014.04.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/23/2014] [Accepted: 04/28/2014] [Indexed: 12/26/2022]
Abstract
The liver has a remarkable capacity to regenerate after injury. Although the regulatory mechanisms of hepatocytic regeneration have been a subject of intense study, the dynamism of the sinusoids, specialized blood vessels in the liver, remains largely unknown. Transient activation of hepatic stellate cells and hepatic sinusoidal endothelial cells, which constitute the sinusoids, contributes to liver regeneration during acute injury, whereas their sustained activation causes liver fibrosis during chronic injury. We focused on understanding the association between damaged hepatocytes and sinusoidal regeneration or liver fibrogenesis using a carbon tetrachloride-induced liver injury mouse model. Damaged hepatocytes rapidly expressed semaphorin 3E (Sema3e), which induced contraction of sinusoidal endothelial cells and thereby contributed to activating hepatic stellate cells for wound healing. In addition, ectopic and consecutive expression of Sema3e in hepatocytes by the hydrodynamic tail-vein injection method resulted in disorganized regeneration of sinusoids and sustained activation of hepatic stellate cells. In contrast, liver fibrosis ameliorated in Sema3e-knockout mice compared with wild-type mice in a chronic liver injury model. Our results indicate that Sema3e, secreted by damaged hepatocytes, affects sinusoidal regeneration in a paracrine manner during liver regeneration, suggesting that Sema3e is a novel therapeutic target in liver fibrogenesis.
Collapse
Affiliation(s)
- Tomoki Yagai
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Minoru Tanaka
- Laboratory of Stem Cell Regulation, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan; Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan.
| |
Collapse
|
177
|
Zigmond E, Samia-Grinberg S, Pasmanik-Chor M, Brazowski E, Shibolet O, Halpern Z, Varol C. Infiltrating Monocyte-Derived Macrophages and Resident Kupffer Cells Display Different Ontogeny and Functions in Acute Liver Injury. THE JOURNAL OF IMMUNOLOGY 2014; 193:344-53. [DOI: 10.4049/jimmunol.1400574] [Citation(s) in RCA: 305] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
178
|
Horwitz E, Stein I, Andreozzi M, Nemeth J, Shoham A, Pappo O, Schweitzer N, Tornillo L, Kanarek N, Quagliata L, Zreik F, Porat RM, Finkelstein R, Reuter H, Koschny R, Ganten T, Mogler C, Shibolet O, Hess J, Breuhahn K, Grunewald M, Schirmacher P, Vogel A, Terracciano L, Angel P, Ben-Neriah Y, Pikarsky E. Human and mouse VEGFA-amplified hepatocellular carcinomas are highly sensitive to sorafenib treatment. Cancer Discov 2014; 4:730-43. [PMID: 24687604 DOI: 10.1158/2159-8290.cd-13-0782] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Death rates from hepatocellular carcinoma (HCC) are steadily increasing, yet therapeutic options for advanced HCC are limited. We identify a subset of mouse and human HCCs harboring VEGFA genomic amplification, displaying distinct biologic characteristics. Unlike common tumor amplifications, this one seems to work via heterotypic paracrine interactions; stromal VEGF receptors (VEGFR), responding to tumor VEGF-A, produce hepatocyte growth factor (HGF) that reciprocally affects tumor cells. VEGF-A inhibition results in HGF downregulation and reduced proliferation, specifically in amplicon-positive mouse HCCs. Sorafenib-the first-line drug in advanced HCC-targets multiple kinases, including VEGFRs, but has only an overall mild beneficial effect. We found that VEGFA amplification specifies mouse and human HCCs that are distinctly sensitive to sorafenib. FISH analysis of a retrospective patient cohort showed markedly improved survival of sorafenib-treated patients with VEGFA-amplified HCCs, suggesting that VEGFA amplification is a potential biomarker for HCC response to VEGF-A-blocking drugs. SIGNIFICANCE Using a mouse model of inflammation-driven cancer, we identified a subclass of HCC carrying VEGFA amplification, which is particularly sensitive to VEGF-A inhibition. We found that a similar amplification in human HCC identifies patients who favorably responded to sorafenib-the first-line treatment of advanced HCC-which has an overall moderate therapeutic efficacy.
Collapse
Affiliation(s)
- Elad Horwitz
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ilan Stein
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mariacarla Andreozzi
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Julia Nemeth
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Avivit Shoham
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Orit Pappo
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Nora Schweitzer
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luigi Tornillo
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Naama Kanarek
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luca Quagliata
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Farid Zreik
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Rinnat M Porat
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Rutie Finkelstein
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hendrik Reuter
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ronald Koschny
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tom Ganten
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Carolin Mogler
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Oren Shibolet
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jochen Hess
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University
| | - Kai Breuhahn
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Myriam Grunewald
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Peter Schirmacher
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Arndt Vogel
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luigi Terracciano
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Peter Angel
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Yinon Ben-Neriah
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Eli Pikarsky
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
179
|
Matsumoto K, Ema M. Roles of VEGF-A signalling in development, regeneration, and tumours. J Biochem 2014; 156:1-10. [DOI: 10.1093/jb/mvu031] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
180
|
Highly robust protein production by co-culture of CHO spheroids layered on feeder cells in serum-free medium. Colloid Polym Sci 2014. [DOI: 10.1007/s00396-013-3093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
181
|
Canavese M, Spaccapelo R. Protective or pathogenic effects of vascular endothelial growth factor (VEGF) as potential biomarker in cerebral malaria. Pathog Glob Health 2014; 108:67-75. [PMID: 24601908 DOI: 10.1179/2047773214y.0000000130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cerebral malaria (CM) is the major lethal complication of Plasmodium falciparum infection. It is characterized by persistent coma along with symmetrical motor signs. Several clinical, histopathological, and laboratory studies have suggested that cytoadherence of parasitized erythrocytes, neural injury by malarial toxin, and excessive inflammatory cytokine production are possible pathogenic mechanisms. Although the detailed pathophysiology of CM remains unsolved, it is thought that the binding of parasitized erythrocytes to the cerebral endothelia of microvessels, leading to their occlusion and the consequent angiogenic dysregulation play a key role in the disease pathogenesis. Recent evidences showed that vascular endothelial growth factor (VEGF) and its receptor-related molecules are over-expressed in the brain tissues of CM patients, as well as increased levels of VEGF are detectable in biologic samples from malaria patients. Whether the modulation of VEGF is causative agent of CM mortality or a specific phenotype of patients with susceptibility to fatal CM needs further evaluation. Currently, there is no biological test available to confirm the diagnosis of CM and its complications. It is hoped that development of biomarkers to identify patients and potential risk for adverse outcomes would greatly enhance better intervention and clinical management to improve the outcomes. We review and discuss here what it is currently known in regard to the role of VEGF in CM as well as VEGF as a potential biomarker.
Collapse
|
182
|
Hu J, Srivastava K, Wieland M, Runge A, Mogler C, Besemfelder E, Terhardt D, Vogel MJ, Cao L, Korn C, Bartels S, Thomas M, Augustin HG. Endothelial cell-derived angiopoietin-2 controls liver regeneration as a spatiotemporal rheostat. Science 2014; 343:416-9. [PMID: 24458641 DOI: 10.1126/science.1244880] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Liver regeneration requires spatially and temporally precisely coordinated proliferation of the two major hepatic cell populations, hepatocytes and liver sinusoidal endothelial cells (LSECs), to reconstitute liver structure and function. The underlying mechanisms of this complex molecular cross-talk remain elusive. Here, we show that the expression of Angiopoietin-2 (Ang2) in LSECs is dynamically regulated after partial hepatectomy. During the early inductive phase of liver regeneration, Ang2 down-regulation leads to reduced LSEC transforming growth factor-β1 production, enabling hepatocyte proliferation by releasing an angiocrine proliferative brake. During the later angiogenic phase of liver regeneration, recovery of endothelial Ang2 expression enables regenerative angiogenesis by controlling LSEC vascular endothelial growth factor receptor 2 expression. The data establish LSECs as a dynamic rheostat of liver regeneration, spatiotemporally orchestrating hepatocyte and LSEC proliferation through angiocrine- and autocrine-acting Ang2, respectively.
Collapse
Affiliation(s)
- Junhao Hu
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), DKFZ-Center for Molecular Biology Alliance, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Gerold G, Pietschmann T. A circuit of paracrine signals between liver sinusoid endothelial cells and hepatocytes regulates hepatitis C virus replication. Hepatology 2014; 59:363-5. [PMID: 23857467 DOI: 10.1002/hep.26621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 07/01/2013] [Accepted: 07/01/2013] [Indexed: 12/07/2022]
Affiliation(s)
- Gisa Gerold
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research, Hannover, Germany
| | | |
Collapse
|
184
|
Bahde R, Kapoor S, Gupta S. Nonselective inhibition of prostaglandin-endoperoxide synthases by naproxen ameliorates acute or chronic liver injury in animals. Exp Mol Pathol 2014; 96:27-35. [PMID: 24220607 PMCID: PMC3944912 DOI: 10.1016/j.yexmp.2013.10.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/31/2013] [Indexed: 12/14/2022]
Abstract
The rising prevalence of hepatic injury due to toxins, metabolites, viruses, etc., necessitates development of further mechanisms for protecting the liver and for treating acute or chronic liver diseases. To examine whether inhibition of inflammation is directed by cyclo-oxygenase pathways, we performed animal studies with naproxen, which inhibits prostaglandin-endoperoxide synthases 1 and 2 and is in extensive clinical use. We administered carbon tetrachloride to induce acute liver injury and ligated the common bile duct to induce chronic liver injury in adult rats. These experimental manipulations produced abnormalities in liver tests, tissue necrosis, compensatory hepatocyte or biliary proliferation, and onset of fibrosis, particularly after bile duct ligation. After carbon tetrachloride-induced acute injury, naproxen decreased liver test abnormalities, tissue necrosis and compensatory hepatocellular proliferation. After bile duct ligation-induced chronic injury, naproxen decreased liver test abnormalities, tissue injury and compensatory biliary hyperplasia. Moreover, after bile duct ligation, naproxen-treated rats showed more periductular oval liver cells, which have been classified as hepatic progenitor cells. In naproxen-treated rats, we found greater expression in hepatic stellate cells and mononuclear cells of cytoprotective factors, such as vascular endothelial growth factor. The ability of naproxen to induce expression of vascular endothelial growth factor was verified in cell culture studies with CFSC-8B clone of rat hepatic stellate cells. Whereas assays for carbon tetrachloride toxicity using cultured primary hepatocytes established that naproxen was not directly cytoprotective, we found conditioned medium containing vascular endothelial growth factor from naproxen-treated CFSC-8B cells protected hepatocytes from carbon tetrachloride toxicity. Therefore, naproxen was capable of ameliorating toxic liver injury, which involved naproxen-induced release of physiological cytoprotective factors in nonparenchymal liver cells. Such drug-induced release of endogenous cytoprotectants will advance therapeutic development for hepatic injury.
Collapse
Affiliation(s)
- Ralf Bahde
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Surgical Research, Department of General and Visceral Surgery, University Hospital, Muenster, Germany
| | - Sorabh Kapoor
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pathology, Marion Bessin Liver Research Center, Diabetes Center, Cancer Center, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, and Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
185
|
Rowe IA, Galsinh SK, Wilson GK, Parker R, Durant S, Lazar C, Branza-Nichita N, Bicknell R, Adams DH, Balfe P, McKeating JA. Paracrine signals from liver sinusoidal endothelium regulate hepatitis C virus replication. Hepatology 2014; 59:375-84. [PMID: 23775568 PMCID: PMC3992845 DOI: 10.1002/hep.26571] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 05/31/2013] [Indexed: 12/15/2022]
Abstract
UNLABELLED Hepatitis C virus (HCV) is a major cause of global morbidity, causing chronic liver injury that can progress to cirrhosis and hepatocellular carcinoma. The liver is a large and complex organ containing multiple cell types, including hepatocytes, sinusoidal endothelial cells (LSEC), Kupffer cells, and biliary epithelial cells. Hepatocytes are the major reservoir supporting HCV replication; however, the role of nonparenchymal cells in the viral lifecycle remains largely unexplored. LSEC secrete factors that promote HCV infection and transcript analysis identified bone morphogenetic protein 4 (BMP4) as a candidate endothelial-expressed proviral molecule. Recombinant BMP4 increased HCV replication and neutralization of BMP4 abrogated the proviral activity of LSEC-conditioned media. Importantly, BMP4 expression was negatively regulated by vascular endothelial growth factor A (VEGF-A) by way of a VEGF receptor-2 (VEGFR-2) primed activation of p38 MAPK. Consistent with our in vitro observations, we demonstrate that in normal liver VEGFR-2 is activated and BMP4 expression is suppressed. In contrast, in chronic liver disease including HCV infection where there is marked endothelial cell proliferation, we observed reduced endothelial cell VEGFR-2 activation and a concomitant increase in BMP4 expression. CONCLUSION These studies identify a role for LSEC and BMP4 in HCV infection and highlight BMP4 as a new therapeutic target for treating individuals with liver disease.
Collapse
Affiliation(s)
- Ian A Rowe
- Hepatitis C Virus Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK,Centre for Liver Research and NIHR Birmingham Liver Biomedical Research Unit, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Sukhdeep K Galsinh
- Hepatitis C Virus Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Garrick K Wilson
- Hepatitis C Virus Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Richard Parker
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research Unit, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Sarah Durant
- Angiogenesis Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Catalin Lazar
- Viral Glycoproteins Department, Institute of BiochemistryBucharest, Romania
| | | | - Roy Bicknell
- Angiogenesis Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - David H Adams
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research Unit, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Peter Balfe
- Hepatitis C Virus Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Jane A McKeating
- Hepatitis C Virus Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK,Centre for Liver Research and NIHR Birmingham Liver Biomedical Research Unit, Institute for Biomedical Research, University of BirminghamBirmingham, UK,Address reprint requests to: Professor Jane McKeating, Hepatitis C Virus Research Group, Institute for Biomedical Research, University of Birmingham, Birmingham UK B15 2TT. E-mail: fax: +44 (0)1214143599
| |
Collapse
|
186
|
De Leu N, Heremans Y, Coppens V, Van Gassen N, Cai Y, D'Hoker J, Magenheim J, Salpeter S, Swisa A, Khalaileh A, Arnold C, Gradwohl G, Van de Casteele M, Keshet E, Dor Y, Heimberg H. Short-term overexpression of VEGF-A in mouse beta cells indirectly stimulates their proliferation and protects against diabetes. Diabetologia 2014; 57:140-7. [PMID: 24121626 DOI: 10.1007/s00125-013-3076-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 09/17/2013] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Vascular endothelial growth factor (VEGF) has been recognised by loss-of-function experiments as a pleiotropic factor with importance in embryonic pancreas development and postnatal beta cell function. Chronic, nonconditional overexpression of VEGF-A has a deleterious effect on beta cell development and function. We report, for the first time, a conditional gain-of-function study to evaluate the effect of transient VEGF-A overexpression by adult pancreatic beta cells on islet vasculature and beta cell proliferation and survival, under both normal physiological and injury conditions. METHODS In a transgenicmouse strain, overexpressing VEGF-A in a doxycycline-inducible and beta cell-specific manner, we evaluated the ability of VEGF-A to affect islet vessel density, beta cell proliferation and protection of the adult beta cell mass from toxin-induced injury. RESULTS Short-term VEGF-A overexpression resulted in islet hypervascularisation, increased beta cell proliferation and protection from toxin-mediated beta cell death, and thereby prevented the development of hyperglycaemia. Extended overexpression of VEGF-A led to impaired glucose tolerance, elevated fasting glycaemia and a decreased beta cell mass. CONCLUSIONS/INTERPRETATION Overexpression of VEGF-A in beta cells time-dependently affects glycometabolic control and beta cell protection and proliferation. These data nourish further studies to examine the role of controlled VEGF delivery in (pre)clinical applications aimed at protecting and/or restoring the injured beta cell mass.
Collapse
|
187
|
Radiofrequency ablation suppresses distant tumour growth in a novel rat model of multifocal hepatocellular carcinoma. Clin Sci (Lond) 2013; 126:243-52. [PMID: 23822114 DOI: 10.1042/cs20130089] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RFA (radiofrequency ablation) is an established therapy for HCC (hepatocellular carcinoma). The multikinase inhibitor sorafenib prolongs survival in advanced HCC. We examined the effects of RFA alone and in combination with sorafenib on a bystanding tumour in a two-tumour rat model of HCC. A total of 80 rats were implanted with two liver tumours and randomized to four treatment groups: vehicle and sham operation (control), sorafenib and sham operation (Sora/Sham), vehicle and RFA (Vh/RFA), and sorafenib and RFA (Sora/RFA) (n=10/group per time point). RFA or sham-operation was performed on the left lobe tumour on day 15. Animals were killed at day 18 and day 30. Non-RFA-targeted right lobe tumours were analysed for angiogenesis, growth factors [HGF (hepatocyte growth factor), EGF (epidermal growth factor) and VEGF (vascular endothelial growth factor)] and infiltrating immune cells (CD3 and CD68). At day 30, the non-RFA-targeted tumours were significantly smaller in all three treatment groups compared with control (Sora/Sham P≤0.0001, Vh/RFA P=0.005 and Sora/RFA P≤0.0001). The smallest tumours were observed in animals treated with a combination of sorafenib and RFA, whereas the size reduction seen in the RFA-only group indicated an RFA-mediated distant suppression of tumour growth. Growth factor measurement revealed transiently decreased EGF levels after RFA (P=0.008), whereas sorafenib treatment decreased HGF levels (P=0.001). MVD (microvessel density) was reduced by sorafenib (P=0.002) despite increased VEGF levels (P≤0.0001). The immune parameters revealed augmented T-cells and IL-10 (interleukin 10) levels in all three treatment groups; sorafenib additionally increased macrophage numbers (P≤0.0001). RFA and sorafenib alone resulted in significant volume reduction of the non-RFA-targeted tumour; this effect was enhanced when both modalities were combined.
Collapse
|
188
|
Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis. Nature 2013; 505:97-102. [PMID: 24256728 PMCID: PMC4142699 DOI: 10.1038/nature12681] [Citation(s) in RCA: 486] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
Chemical or traumatic damage to the liver is frequently associated with aberrant healing(fibrosis) that overrides liver regeneration1–5. The mechanism by which hepatic niche cells differentially modulate regeneration and fibrosis during liver repair remains to be defined6–8. Hepatic vascular niche predominantly represented by liver sinusoidal endothelial cells (LSECs), deploys paracrine trophogens, known as angiocrine factors, to stimulate regeneration9–15. Nevertheless, it remains unknown how pro-regenerative angiocrine signals from LSECs is subverted to promote fibrosis16,17. Here, by combining inducible endothelial cell (EC)-specific mouse gene deletion strategy and complementary models of acute and chronic liver injury, we revealed that divergent angiocrine signals from LSECs elicit regeneration after immediateinjury and provoke fibrosis post chronic insult. The pro-fibrotic transition of vascular niche results from differential expression of stromal derived factor-1 (SDF-1) receptors, CXCR7 and CXCR418–21in LSECs. After acute injury, CXCR7 upregulation in LSECs acts in conjunction with CXCR4 to induce transcription factor Id1, deploying pro-regenerative angiocrine factors and triggering regeneration. Inducible deletion of Cxcr7 in adult mouse LSECs (Cxcr7iΔEC/iΔEC) impaired liver regeneration by diminishing Id1-mediated production of angiocrine factors9–11. By contrast, after chronic injury inflicted by iterative hepatotoxin (carbon tetrachloride) injection and bile duct ligation, constitutive FGFR1 signaling in LSECs counterbalanced CXCR7-dependent pro-regenerative response and augmented CXCR4 expression. This predominance of CXCR4 over CXCR7 expression shifted angiocrine response of LSECs, stimulating proliferation of desmin+hepatic stellate-like cells22,23 and enforcing a pro-fibrotic vascular niche. EC-specific ablation of either Fgfr1 (Fgfr1iΔEC/iΔEC) or Cxcr4 (Cxcr4iΔEC/iΔEC) in mice restored pro-regenerative pathway and prevented FGFR1-mediated maladaptive subversion of angiocrine factors. Similarly, selective CXCR7 activation in LSECs abrogated fibrogenesis. Thus, we have demonstrated that in response to liver injury, differential recruitment of pro-regenerative CXCR7/Id1 versus pro-fibrotic FGFR1/CXCR4 angiocrine pathways in vascular niche balances regeneration and fibrosis. These results provide a therapeutic roadmap to achieve hepatic regeneration without provoking fibrosis1,2,4.
Collapse
|
189
|
Abstract
Angiogenesis, defined as the formation of new microvasculature from preexisting blood vessels and mature endothelial cells, plays a major role in wound healing and scar formation, and it is associated with inflammatory responses. Angiogenesis can occur in physiological conditions, such as during liver regeneration, and in pathological situations, such as during the progression of fibrosis to cirrhosis and also during tumor angiogenesis. Cellular cross-talk among liver sinusoidal endothelial cells (LSECs), hepatic stellate cells and hepatocytes is believed to play an important role in the angiogenesis process during both liver regeneration and development of cirrhosis. In addition to mature endothelial cells, bone marrow (BM)-derived circulating endothelial progenitor cells (EPCs) have been recently identified for their contribution to post-natal vasculogenesis/angiogenesis. In vivo, EPCs are mobilized into the peripheral blood in response to tissue ischemia or traumatic injury, migrate to the sites of injured endothelium and differentiate into mature endothelial cells. In our recent studies, we have explored the role of EPC-mediated angiogenesis in liver regeneration and/or cirrhosis. Results have demonstrated significantly increased endogenous levels of circulating EPCs in cirrhotic patients in comparison to the controls. Also, EPCs from cirrhotic patients have been observed to stimulate substantial angiogenesis by resident LSECs in vitro via paracrine factors such as vascular endothelial growth factor and platelet-derived growth factor. This review gives an overview of the angiogenesis process in liver regeneration and disease and discusses a new mechanism for intrahepatic angiogenesis mediated by BM-derived EPCs.
Collapse
Affiliation(s)
- Savneet Kaur
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, UP, India.
| | - K Anita
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, UP, India
| |
Collapse
|
190
|
Fomin ME, Zhou Y, Beyer AI, Publicover J, Baron JL, Muench MO. Production of factor VIII by human liver sinusoidal endothelial cells transplanted in immunodeficient uPA mice. PLoS One 2013; 8:e77255. [PMID: 24167566 PMCID: PMC3805584 DOI: 10.1371/journal.pone.0077255] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 09/02/2013] [Indexed: 12/23/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) form a semi-permeable barrier between parenchymal hepatocytes and the blood. LSECs participate in liver metabolism, clearance of pathological agents, immunological responses, architectural maintenance of the liver and synthesis of growth factors and cytokines. LSECs also play an important role in coagulation through the synthesis of Factor VIII (FVIII). Herein, we phenotypically define human LSECs isolated from fetal liver using flow cytometry and immunofluorescence microscopy. Isolated LSECs were cultured and shown to express endothelial markers and markers specific for the LSEC lineage. LSECs were also shown to engraft the liver when human fetal liver cells were transplanted into immunodeficient mice with liver specific expression of the urokinase-type plasminogen activator (uPA) transgene (uPA-NOG mice). Engrafted cells expressed human Factor VIII at levels approaching those found in human plasma. We also demonstrate engraftment of adult LSECs, as well as hepatocytes, transplanted into uPA-NOG mice. We propose that overexpression of uPA provides beneficial conditions for LSEC engraftment due to elevated expression of the angiogenic cytokine, vascular endothelial growth factor. This work provides a detailed characterization of human midgestation LSECs, thereby providing the means for their purification and culture based on their expression of CD14 and CD32 as well as a lack of CD45 expression. The uPA-NOG mouse is shown to be a permissive host for human LSECs and adult hepatocytes, but not fetal hepatoblasts. Thus, these mice provide a useful model system to study these cell types in vivo. Demonstration of human FVIII production by transplanted LSECs encourages further pursuit of LSEC transplantation as a cellular therapy for the treatment of hemophilia A.
Collapse
Affiliation(s)
- Marina E. Fomin
- Blood Systems Research Institute, San Francisco, California, United States of America
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Yanchen Zhou
- Blood Systems Research Institute, San Francisco, California, United States of America
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Ashley I. Beyer
- Blood Systems Research Institute, San Francisco, California, United States of America
| | - Jean Publicover
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Liver Center, University of California San Francisco, San Francisco, California, United States of America
| | - Jody L. Baron
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Liver Center, University of California San Francisco, San Francisco, California, United States of America
| | - Marcus O. Muench
- Blood Systems Research Institute, San Francisco, California, United States of America
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
- Liver Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
191
|
Moreno-Carranza B, Goya-Arce M, Vega C, Adán N, Triebel J, López-Barrera F, Quintanar-Stéphano A, Binart N, Martínez de la Escalera G, Clapp C. Prolactin promotes normal liver growth, survival, and regeneration in rodents: effects on hepatic IL-6, suppressor of cytokine signaling-3, and angiogenesis. Am J Physiol Regul Integr Comp Physiol 2013; 305:R720-6. [DOI: 10.1152/ajpregu.00282.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Prolactin (PRL) is a potent liver mitogen and proangiogenic hormone. Here, we used hyperprolactinemic rats and PRL receptor-null mice (PRLR−/−) to study the effect of PRL on liver growth and angiogenesis before and after partial hepatectomy (PH). Liver-to-body weight ratio (LBW), hepatocyte and sinusoidal endothelial cell (SEC) proliferation, and hepatic expression of VEGF were measured before and after PH in hyperprolactinemic rats, generated by placing two anterior pituitary glands (AP) under the kidney capsule. Also, LBW and hepatic expression of IL-6, as well as suppressor of cytokine signaling-3 (SOCS-3), were evaluated in wild-type and PRLR−/−mice before and after PH. Hyperprolactinemia increased the LBW, the proliferation of hepatocytes and SECs, and VEGF hepatic expression. Also, liver regeneration was increased in AP-grafted rats and was accompanied by elevated hepatocyte and SEC proliferation, and VEGF expression compared with nongrafted controls. Lowering circulating PRL levels with CB-154, an inhibitor of AP PRL secretion, prevented AP-induced stimulation of liver growth. Relative to wild-type animals, PRLR−/−mice had smaller livers, and soon after PH, they displayed an approximately twofold increased mortality and elevated and reduced hepatic IL-6 and SOCS-3 expression, respectively. However, liver regeneration was improved in surviving PRLR−/−mice. PRL stimulates normal liver growth, promotes survival, and regulates liver regeneration by mechanisms that may include hepatic downregulation of IL-6 and upregulation of SOCS-3, increased hepatocyte proliferation, and angiogenesis. PRL contributes to physiological liver growth and has potential clinical utility for ensuring survival and regulating liver mass in diseases, injuries, or surgery of the liver.
Collapse
Affiliation(s)
| | - Maite Goya-Arce
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Claudia Vega
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Norma Adán
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Jakob Triebel
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | | | - Andrés Quintanar-Stéphano
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México; and
| | - Nadine Binart
- Institut National de la Santé et de la Recherche Médicale, U693, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
192
|
Abstract
Vascular endothelial growth factor receptors (VEGFRs) in vertebrates play essential roles in the regulation of angiogenesis and lymphangiogenesis. VEGFRs belong to the receptor-type tyrosine kinase (RTK) supergene family. They consist of a ligand-binding region with seven immunoglobulin (7 Ig) -like domains, a trans-membrane (TM) domain, and a tyrosine kinase (TK) domain with a long kinase insert (KI) (also known as a type-V RTK). Structurally, VEGFRs are distantly related to the members of the M-colony stimulating factor receptor/platelet-derived growth factor receptor (CSFR)/(PDGFR) family, which have five immunoglobulin (5 Ig)-like domains. However, signal transduction in VEGFRs significantly differs from that in M-CSFR/PDGFRs. VEGFR2, the major signal transducer for angiogenesis, preferentially uses the phospholipase Cγ-protein kinase C (PLC-γ-PKC)-MAPK pathway, whereas M-CSFR/PDGFRs use the PI3 kinase-Ras-MAPK pathway for cell proliferation. In phylogenetic development, the VEGFR-like receptor in nonvertebrates appears to be the ancestor of the 7 Ig- and 5 Ig-RTK families because most nonvertebrates have only a single 7 Ig-RTK gene. In mammals, VEGFRs are deeply involved in pathological angiogenesis, including cancer and inflammation. Thus, an efficient inhibitor targeting VEGFRs could be useful in suppressing various diseases.
Collapse
|
193
|
Chatziioannou AN, Siskos AP, Loxas D, Kavatzas N, Agrogiannis G, Fokas D, Malagari K, Kostomitsopoulos NG, Tsigkou O, Tamvakopoulos C. Transarterial embolization with sorafenib in animal livers: a pharmacokinetics study. J Vasc Interv Radiol 2013; 24:1657-63.e1. [PMID: 24060438 DOI: 10.1016/j.jvir.2013.08.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 08/13/2013] [Accepted: 08/13/2013] [Indexed: 10/26/2022] Open
Abstract
PURPOSE To assess the safety and feasibility of the targeted delivery of the antiangiogenic drug sorafenib to the liver using transarterial chemoembolization methodology as a novel approach to hepatocellular carcinoma (HCC) therapy. MATERIALS AND METHODS Seven healthy New Zealand white rabbits were used in the study. After placement of a catheter in the common hepatic artery, six rabbits were treated with chemoembolization of sorafenib in iodized oil (Lipiodol) (sorafenib dose 0.1 mg/kg), and one rabbit received Lipiodol only. Liquid chromatography tandem mass spectrometry was used to measure the concentration of sorafenib in the peripheral blood and liver tissue 24 hours and 72 hours after treatment. Histochemical staining of the liver sections and biochemical measurements were performed. RESULTS The administration of sorafenib in Lipiodol emulsions by transarterial chemoembolization resulted in sorafenib concentrations of 794 ng/g ± 240 and 64 ng/g ± 15 in the liver tissue 24 hours and 72 hours after treatment. The average liver-to-serum ratios 24 hours and 72 hours after treatment were approximately 14 and 22. The histochemical staining of the liver tissue sections and aspartate aminotransferase, alanine aminotransferase, γ-glutamyltransferase and total bilirubin concentrations indicated no significant liver damage. CONCLUSIONS Transarterial chemoembolization with sorafenib in Lipiodol is an effective methodology for the localized delivery of this drug to the liver and has possible practical implications in therapeutic interventions for the treatment of hepatocellular carcinoma.
Collapse
|
194
|
Wei R, Yang J, Hong TP. Relationship between vascular endothelial cells and pancreatic islet development and stem cell differentiation. Shijie Huaren Xiaohua Zazhi 2013; 21:2493-2499. [DOI: 10.11569/wcjd.v21.i25.2493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As the main components of the pancreatic islet niche, endothelial cells participate in many processes of pancreatic development, including pancreatic cell fate decision, endocrine pancreatic cell differentiation and proliferation, and spatial distribution of the pancreas. On different occasions, endothelial cells play disparate roles by cross-talking with islet cells to influence endocrine pancreatic cell differentiation and islet morphology and function. Cytokines such as hepatocyte growth factor and sphingosine-1-phosphate as well as the extracellular matrixes such as laminin and collagen Ⅳ, which are produced and/or secreted by endothelial cells, play important roles in the regulation of islet development and function. Furthermore, endothelial cells are involved in the balance between self-renewal and differentiation of stem cells. Application of endothelial cells to induce the differentiation of stem cells into functional islet cells may be one of the most promising approaches to cell replacement therapy for diabetes.
Collapse
|
195
|
Stöppeler S, Palmes D, Fehr M, Hölzen JP, Zibert A, Siaj R, Schmidt HHJ, Spiegel HU, Bahde R. Gender and strain-specific differences in the development of steatosis in rats. Lab Anim 2013; 47:43-52. [PMID: 23467489 DOI: 10.1177/0023677212473717] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common problem with a wide variety of phenotypes. While its pathogenesis is still not fully understood, several risk factors for disease progression have been identified. Therefore, defining adequate animal models may serve to unreveal the pathogenesis in NAFLD. We studied Lewis and Sprague-Dawley rats of both genders (n = 6) fed standard (Std) or high-fat (HF) diet for three weeks. Disease stage was assessed by haematoxylin-eosin, Azan Heidenheim and Oil-Red staining, apoptosis by single-stranded DNA (ssDNA) detection and liver regeneration by Ki-67 staining. Serum markers of liver injury and lipid metabolism including adipocytokines were analysed. Livers of both strains and genders fed with HF diet demonstrated evidence of steatosis. Lewis rats developed microvesicular steatosis whereas Sprague-Dawley rats presented macrovesicular steatosis accompanied by pronounced fibrosis. Female gender of both strains was associated with lower steatosis grade and higher proliferation rate (P < 0.05). Gender-specific differences were most prominent in Lewis rats on a HF diet, where females showed lower alkaline phosphatase, cholesterol, triglyceride and leptin levels and a more favourable low-density lipoprotein/high-density lipoprotein ratio than males (P < 0.05). Reverse transcriptase-polymerase chain reaction analysis was performed to demonstrate changes in expression of various genes important for liver regeneration, fibrosis and steatosis. HF diet induced downregulation of proangiogenic genes such as vascular endothelial growth factor receptor 1 and 2 (P < 0.05) in males was not present in females. In conclusion, strain and gender served major roles in disease progression. These differences should be considered when designing studies and may offer new ways to advance therapeutic strategies.
Collapse
Affiliation(s)
- S Stöppeler
- Surgical Research, Department of General and Visceral Surgery, Muenster University Hospital, Muenster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Cogger VC, Roessner U, Warren A, Fraser R, Le Couteur DG. A Sieve-Raft Hypothesis for the regulation of endothelial fenestrations. Comput Struct Biotechnol J 2013; 8:e201308003. [PMID: 24688743 PMCID: PMC3962122 DOI: 10.5936/csbj.201308003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 07/31/2013] [Accepted: 08/11/2013] [Indexed: 01/13/2023] Open
Affiliation(s)
- Victoria C Cogger
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| | - Ute Roessner
- Metabolomics Australia and Australian Centre for Plant Functional Genomics, The University of Melbourne, 3010 Victoria, Australia
| | - Alessandra Warren
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| | - Robin Fraser
- Christchurch School of Medicine, University of Otago, Christchurch NZ
| | - David G Le Couteur
- Centre for Education and Research on Ageing and ANZAC Research Institute, Concord Hospital and University of Sydney, Sydney NSW, Australia ; Charles Perkins Centre, University of Sydney NSW Australia
| |
Collapse
|
197
|
Nejak-Bowen KN, Orr AV, Bowen WC, Michalopoulos GK. Gliotoxin-induced changes in rat liver regeneration after partial hepatectomy. Liver Int 2013; 33:1044-1055. [PMID: 23552057 PMCID: PMC3706483 DOI: 10.1111/liv.12164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 03/10/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND Hepatic non-parenchymal cells (NPCs), encompassing hepatic stellate cells (HSCs), macrophages and endothelial cells, synthesize new hepatocyte growth factor (HGF) during liver regeneration (LR), and also play an important function in matrix production at the end of regeneration. AIMS The aim of this study was to determine whether ablating NPCs either during hepatocyte proliferation or during matrix resynthesis will have any effect on LR. METHODS Rats were injected with either gliotoxin (which induces NPC apoptosis) or vehicle control at various stages during partial hepatectomy (PH). NPCs and hepatocytes were also treated in vitro with gliotoxin. RESULTS Proliferating cells were abundant in control livers 24 h after PH, while in gliotoxin-treated rats, mitosis was absent, apoptotic NPCs were apparent and HGF was decreased. In vitro studies demonstrated a > 50% decrease in cell viability in NPC cultures, while hepatocyte viability and proliferation were unaffected. Chronic elimination of NPCs over a period of 5 days after PH led to increased desmin-positive HSCs and fewer alpha smooth muscle actin-expressing HSCs. Finally, there was continued proliferation of hepatocytes and decreased collagen I and TGF-β when HSCs, the matrix-producing NPCs, were ablated during later stages of LR. CONCLUSIONS Ablation of NPCs at early time points after PH interferes with liver regeneration, while their ablation at late stages causes impairment in the termination of LR, demonstrating a time-dependent regulatory role of NPCs in the regenerative process.
Collapse
|
198
|
Kapadia S, Hapani S, Choueiri TK, Wu S. Risk of liver toxicity with the angiogenesis inhibitor pazopanib in cancer patients. Acta Oncol 2013; 52:1202-12. [PMID: 23594201 DOI: 10.3109/0284186x.2013.782103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE The angiogenesis inhibitor pazopanib has been approved for the treatment of advanced renal cell cancer (RCC) and soft tissue sarcoma. Severe and fatal hepatotoxicity has been observed in its clinical studies. This analysis was conducted to determine the risk of liver toxicity by a systematic review and meta-analysis of clinical trials. PATIENTS AND METHODS Databases from PubMed, Web of Science and abstracts presented at ASCO meetings up to January, 2012 were searched to identify relevant studies. Eligible studies included prospective trials of cancer patients treated with pazopanib starting at 800 mg daily. Summary incidence rates, relative risks, and 95% confidence intervals (CIs) were calculated using a fixed- or random-effects model. RESULTS A total of 1478 patients from 10 clinical trials were included. The incidences of all-grade aspartate aminotransferase (AST), alanine transaminase (ALT), and bilirubin elevation were 39.6% (95% CI 31.2-48.6%), 41.4% (95% CI 34.1-49.0%), and 24.8% (95% CI 16.3-35.3%), respectively. The incidences of high-grade (Grade 3 and 4) AST, ALT and bilirubin elevation were 6.9% (95% CI 5.5-8.6%), 9.4% (95% CI 7.8-11.4%), and 3.4% (2.4-5.0%), respectively. In comparison with placebo, pazopanib significantly increased the risk of high-grade AST elevation (RR 6.56, 95% CI 2.04-21.07, p = 0.002) and ALT elevation (RR 4.33, 95% CI 1.88-10.0, p = 0.001). However, the risks of high-grade bilirubin elevation (RR 1.31, 95% CI 0.47-3.64) and fatal hepatotoxicity (RR 2.51, 95% CI 0.12-51.91, p = 0.55) were not significantly elevated. CONCLUSION The use of pazopanib was associated with a significantly increased risk of severe non-fatal hepatotoxicity in cancer patients.
Collapse
Affiliation(s)
- Samir Kapadia
- Division of Hematology and Oncology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, New York, USA
| | | | | | | |
Collapse
|
199
|
Abstract
Epoxyeicosatrienoic acids (EETs), lipid mediators produced by cytochrome P450 epoxygenases, regulate inflammation, angiogenesis, and vascular tone. Despite pleiotropic effects on cells, the role of these epoxyeicosanoids in normal organ and tissue regeneration remains unknown. EETs are produced predominantly in the endothelium. Normal organ and tissue regeneration require an active paracrine role of the microvascular endothelium, which in turn depends on angiogenic growth factors. Thus, we hypothesize that endothelial cells stimulate organ and tissue regeneration via production of bioactive EETs. To determine whether endothelial-derived EETs affect physiologic tissue growth in vivo, we used genetic and pharmacological tools to manipulate endogenous EET levels. We show that endothelial-derived EETs play a critical role in accelerating tissue growth in vivo, including liver regeneration, kidney compensatory growth, lung compensatory growth, wound healing, corneal neovascularization, and retinal vascularization. Administration of synthetic EETs recapitulated these results, whereas lowering EET levels, either genetically or pharmacologically, delayed tissue regeneration, demonstrating that pharmacological modulation of EETs can affect normal organ and tissue growth. We also show that soluble epoxide hydrolase inhibitors, which elevate endogenous EET levels, promote liver and lung regeneration. Thus, our observations indicate a central role for EETs in organ and tissue regeneration and their contribution to tissue homeostasis.
Collapse
|
200
|
Yu J, Yin S, Zhang W, Gao F, Liu Y, Chen Z, Zhang M, He J, Zheng S. Hypoxia preconditioned bone marrow mesenchymal stem cells promote liver regeneration in a rat massive hepatectomy model. Stem Cell Res Ther 2013; 4:83. [PMID: 23856418 PMCID: PMC3854783 DOI: 10.1186/scrt234] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/12/2013] [Indexed: 12/14/2022] Open
Abstract
Introduction Bone marrow mesenchymal stem cells (BMMSCs) have been reported to facilitate liver regeneration after toxic injuries. However, the effect of BMMSCs on liver regeneration after massive hepatectomy is barely studied. Here we explored whether infusion of BMMSCs promotes liver regeneration in a rat massive hepatectomy model. Methods Hypoxia preconditioning was achieved by culturing BMMSCs under a hypoxia environment. Then 85% hepatectomy was performed and hypoxia or normoxia preconditioned BMMSCs were infused into the portal vein. A group of rats received vascular endothelial growth factor (VEGF) neutralizing antibody perioperatively, and underwent 85% hepatectomy and a subsequent infusion of hypoxia preconditioned BMMSCs to verify the role of VEGF in the effects of BMMSCs on liver regeneration. Liver samples were collected and liver regeneration was evaluated postoperatively. Results Hypoxia preconditioning enhanced the expression of VEGF in BMMSCs in vitro. Infusion of BMMSCs promoted proliferation of hepatocytes, reflected by elevated cyclin D1 expression and proliferating cell nuclear antigen-positive hepatocytes. However, BMMSC infusion did not improve the serum albumin level, liver weight/body weight ratio, and survival after operation. Infusion of hypoxia preconditioned BMMSCs significantly elevated cyclin D1, proliferating cell nuclear antigen-positive hepatocytes, liver weight/body weight ratio, and survival compared with normoxia preconditioned BMMSCs, accompanied by an increased serum albumin level. The level of VEGF in liver homogenate was much higher in hypoxia preconditioned BMMSC-treated animals than in other groups. In addition, the perioperative injection of VEGF neutralizing antibody significantly blocked the therapeutic effects of hypoxia preconditioned BMMSCs on liver injury and regeneration in this model. Conclusion Hypoxia preconditioned BMMSCs enhanced liver regeneration after massive hepatectomy in rats, possibly by upregulating the level of VEGF.
Collapse
|